Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
1.
J Diabetes Res ; 2024: 5574968, 2024.
Article in English | MEDLINE | ID: mdl-38800586

ABSTRACT

Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Insulin , Islets of Langerhans Transplantation , Myoblasts, Skeletal , Neovascularization, Physiologic , Animals , Islets of Langerhans Transplantation/methods , Diabetes Mellitus, Experimental/metabolism , Myoblasts, Skeletal/transplantation , Myoblasts, Skeletal/metabolism , Mice , Male , Insulin/metabolism , Hepatocyte Growth Factor/metabolism , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/blood supply , Chemokine CXCL12/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/surgery , Signal Transduction , Insulin Secretion , Cell Differentiation
2.
PLoS One ; 16(2): e0247381, 2021.
Article in English | MEDLINE | ID: mdl-33635873

ABSTRACT

Right ventricular dysfunction is a predictor for worse outcomes in patients with congenital heart disease. Myocardial ischemia is primarily associated with right ventricular dysfunction in patients with congenital heart disease and may be a therapeutic target for right ventricular dysfunction. Previously, autologous skeletal myoblast patch therapy showed an angiogenic effect for left ventricular dysfunction through cytokine paracrine effects; however, its efficacy in right ventricular dysfunction has not been evaluated. Thus, this study aimed to evaluate the angiogenic effect of autologous skeletal myoblast patch therapy and amelioration of metabolic and functional dysfunction, in a pressure-overloaded right heart porcine model. Pulmonary artery stenosis was induced by a vascular occluder in minipigs; after two months, autologous skeletal myoblast patch implantation on the right ventricular free wall was performed (n = 6). The control minipigs underwent a sham operation (n = 6). The autologous skeletal myoblast patch therapy alleviated right ventricular dilatation and ameliorated right ventricular systolic and diastolic dysfunction. 11C-acetate kinetic analysis using positron emission tomography showed improvement in myocardial oxidative metabolism and myocardial flow reserve after cell patch implantation. On histopathology, a higher capillary density and vascular maturity with reduction of myocardial ischemia were observed after patch implantation. Furthermore, analysis of mRNA expression revealed that the angiogenic markers were upregulated, and ischemic markers were downregulated after patch implantation. Thus, autologous skeletal myoblast patch therapy ameliorated metabolic and functional dysfunction in a pressure-overloaded right heart porcine model, by alleviating myocardial ischemia through angiogenesis.


Subject(s)
Myoblasts, Skeletal/transplantation , Myocardial Ischemia/prevention & control , Stenosis, Pulmonary Artery/therapy , Ventricular Dysfunction, Right/prevention & control , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Multidetector Computed Tomography , Myocardial Ischemia/etiology , Myocardial Ischemia/metabolism , Neovascularization, Physiologic , Oxidative Stress , Stenosis, Pulmonary Artery/etiology , Stenosis, Pulmonary Artery/metabolism , Swine , Swine, Miniature , Transplantation, Autologous , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/metabolism
3.
Commun Biol ; 3(1): 182, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313117

ABSTRACT

Understanding the signaling pathways that regulate proliferation and differentiation of muscle progenitors is essential for successful cell transplantation for treatment of Duchenne muscular dystrophy. Here, we report that a γ-secretase inhibitor, DAPT (N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine tertial butyl ester), which inhibits the release of NICD (Notch intercellular domain), promotes the fusion of human muscle progenitors in vitro and improves their engraftment in the tibialis anterior muscle of immune-deficient mice. Gene expression analysis revealed that DAPT severely down-regulates PTGER2, which encodes prostaglandin (PG) E2 receptor 2 (EP2), in human muscle progenitors in the differentiation condition. Functional analysis suggested that Notch signaling inhibits differentiation and promotes self-renewal of human muscle progenitors via PGE2/EP2 signaling in a cAMP/PKA-independent manner.


Subject(s)
Cell Differentiation , Cell Proliferation , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Receptor, Notch3/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Differentiation/drug effects , Cell Fusion , Cell Line , Cell Proliferation/drug effects , Cell Self Renewal , Cell Survival , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Dipeptides/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Male , Mice, Inbred NOD , Mice, Inbred mdx , Mice, SCID , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscle, Skeletal/surgery , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/transplantation , Receptor, Notch3/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Signal Transduction , Stem Cell Transplantation
4.
Int J Mol Sci ; 20(21)2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31683627

ABSTRACT

Muscular dystrophies (MD) are a group of genetic diseases that lead to skeletal muscle wasting and may affect many organs (multisystem). Unfortunately, no curative therapies are available at present for MD patients, and current treatments mainly address the symptoms. Thus, stem-cell-based therapies may present hope for improvement of life quality and expectancy. Different stem cell types lead to skeletal muscle regeneration and they have potential to be used for cellular therapies, although with several limitations. In this review, we propose a combination of genetic, biochemical, and cell culture treatments to correct pathogenic genetic alterations and to increase proliferation, dispersion, fusion, and differentiation into new or hybrid myotubes. These boosted stem cells can also be injected into pretreate recipient muscles to improve engraftment. We believe that this combination of treatments targeting the limitations of stem-cell-based therapies may result in safer and more efficient therapies for MD patients. Matricryptins have also discussed.


Subject(s)
Muscular Dystrophies/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cell Differentiation , Humans , Muscular Dystrophies/physiopathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/transplantation , Regeneration , Tissue Engineering/methods
5.
J Neuropathol Exp Neurol ; 78(1): 38-46, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30481300

ABSTRACT

We have previously studied in nonhuman primates several aspects of the acute rejection of myofibers, including the histological characteristics, the mechanisms of myofiber elimination by the T cells, and the development of anti-donor antibodies. Here, we report the participation of the complement membrane attack complex (MAC) in this context. We used muscle sections of macaques from experiments of allogeneic muscle precursor cell transplantation with confirmed rejection of the graft-derived myofibers. Sections were stained with hematoxylin and eosin, alizarin red and for immunodetection of MAC, CD8, CD4, C3, C4d, and immunoglobulins. The prominent finding was the presence of sarcolemmal MAC (sMAC) deposits in biopsies with ongoing acute rejection or with recent acute rejection. The numbers of sMAC-positive myofibers were variable, being higher when there was an intense lymphocyte infiltration. Few sMAC-positive myofibers were necrotic or had evidence of sarcolemma permeation. The immunodetection of C3, C4d, and immunoglobulins did not provide significant elements. In conclusion, sMAC deposits were related to myofiber rejection. The fact that the vast majority of sMAC-positive myofibers had no signs of necrosis or sarcolemmal permeation suggests that MAC would not be harmful to myofibers by itself.


Subject(s)
Complement Membrane Attack Complex/immunology , Graft Rejection/immunology , Myoblasts, Skeletal/transplantation , Sarcolemma/immunology , Animals , Macaca fascicularis
6.
Biomed Pharmacother ; 109: 304-313, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30396088

ABSTRACT

Heart Diseases are serious and global public health concern. In spite of remarkable therapeutic developments, the prediction of patients with Heart Failure (HF) is weak, and present therapeutic attitudes do not report the fundamental problem of the cardiac tissue loss. Innovative therapies are required to reduce mortality and limit or abolish the necessity for cardiac transplantation. Stem cell-based therapies applied to the treatment of heart disease is according to the understanding that natural self-renewing procedures are inherent to the myocardium, nonetheless may not be adequate to recover the infarcted heart muscle. Following the first account of cell therapy in heart diseases, examination has kept up to rapidity; besides, several animals and human clinical trials have been conducted to preserve the capacity of numerous stem cell population in advance cardiac function and decrease infarct size. The purpose of this study was to censoriously evaluate the works performed regarding the usage of four major subgroups of stem cells, including induced Pluripotent Stem Cells (iPSC), Embryonic Stem Cells (ESCs), Cardiac Stem Cells (CDC), and Skeletal Myoblasts, in heart diseases, at the preclinical and clinical studies. Moreover, it is aimed to argue the existing disagreements, unsolved problems, and prospect directions.


Subject(s)
Embryonic Stem Cells/transplantation , Heart Diseases/therapy , Induced Pluripotent Stem Cells/transplantation , Myoblasts, Skeletal/transplantation , Myocytes, Cardiac/transplantation , Stem Cell Transplantation/methods , Animals , Embryonic Stem Cells/metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Myoblasts, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Stem Cell Transplantation/trends
7.
Acta Biomater ; 84: 180-193, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30528606

ABSTRACT

Injectable conductive hydrogels have great potential as tissue engineering scaffolds and delivery vehicles for electrical signal sensitive cell therapy. In this work, we present the synthesis of a series of injectable electroactive degradable hydrogels with rapid self-healing ability and their potential application as cell delivery vehicles for skeletal muscle regeneration. Self-healable conductive injectable hydrogels based on dextran-graft-aniline tetramer-graft-4-formylbenzoic acid and N-carboxyethyl chitosan were synthesized at physiological conditions. The dynamic Schiff base bonds between the formylbenzoic acid and amine group from N-carboxyethyl chitosan endowed the hydrogels with rapid self-healing ability, which was verified by rheological test. Equilibrated swelling ratio, morphology, mechanical strength, electrochemistry and conductivity of the injectable hydrogels were fully investigated. The self-healable conductive hydrogels showed an in vivo injectability and a linear-like degradation behavior. Two different kinds of cells (C2C12 myoblasts and human umbilical vein endothelial cells (HUVEC)) were encapsulated in the hydrogels by self-healing effect. The L929 fibroblast cell culture results indicated the biocompatibility of the hydrogels. Moreover, the C2C12 myoblast cells were released from the conductive hydrogels with a linear-like profile. The in vivo skeletal muscle regeneration was also studied in a volumetric muscle loss injury model. All these data indicated that these biodegradable self-healing conductive hydrogels are potential candidates as cell delivery vehicles and scaffolds for skeletal muscle repair. STATEMENT OF SIGNIFICANCE: Injectable hydrogels with self-healing and electrical conductivity properties are excellent candidates as tissue-engineered scaffolds for myoblast cell therapy and skeletal muscle regeneration. The self-healing property of these hydrogels can prolong their lifespan. However, most of the reported conductive hydrogels are not degradable or do not have the self-healing ability. Herein, we synthesized antibacterial conductive self-healing hydrogels as a cell delivery carrier for cardiac cell therapy based on chitosan-grafted-tetraaniline hydrogels synthesized in our previous work. However, an acid solution was used to dissolve the polymers in that study, which may induce toxicity to cells. In this work, we synthesized a series of injectable electroactive biodegradable hydrogels with rapid self-healing ability composed of N-carboxyethyl chitosan (CECS) and dextran-graft-aniline oligomers, and these hydrogel precusor can dissolve in PBS solution of pH 7.4; we further demonstrated their potential application as cell delivery vehicles for skeletal muscle regeneration.


Subject(s)
Cells, Immobilized , Chitosan , Dextrans , Hydrogels , Muscle, Skeletal/physiology , Myoblasts, Skeletal , Regeneration , Animals , Cell Line , Cells, Immobilized/metabolism , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Chitosan/chemical synthesis , Chitosan/chemistry , Chitosan/pharmacology , Dextrans/chemical synthesis , Dextrans/chemistry , Dextrans/pharmacology , Hydrogels/chemical synthesis , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Muscle, Skeletal/pathology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Myoblasts, Skeletal/transplantation , Rats , Rats, Sprague-Dawley
8.
Stem Cell Res Ther ; 9(1): 316, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30463597

ABSTRACT

BACKGROUND: Human muscle-derived stem cells (hMDSCs) have been shown to regenerate bone efficiently when they were transduced with Lenti-viral bone morphogenetic protein 2 (LBMP2). However, whether the age of hMDSCs and the animal host affect the bone regeneration capacity of hMDSCs and mechanism are unknown which prompted the current study. METHODS: We isolated three gender-matched young and old populations of skeletal muscle stem cells, and tested the influence of cells' age on in vitro osteogenic differentiation using pellet culture before and after Lenti-BMP2/green fluorescent protein (GFP) transduction. We further investigated effects of the age of hMDSCs and animal host on hMDSC-mediated bone regeneration in a critical-size calvarial bone defect model in vivo. Micro-computer tomography (CT), histology, and immunohistochemistry were used to evaluate osteogenic differentiation and mineralization in vitro and bone regeneration in vivo. Western blot, quantitative polymerase chain reaction (PCR), and oxidative stress assay were performed to detect the effects of age of hMDSCs on cell survival and osteogenic-related genes. Serum insulin-like growth factor 1 (IGF1) and receptor activator of nuclear factor-kappa B ligand (RANKL) were measured with an enzyme-linked immunosorbent assay (ELISA). RESULTS: We found LBMP2/GFP transduction significantly enhanced osteogenic differentiation of hMDSCs in vitro, regardless of donor age. We also found old were as efficient as young LBMP2/GFP-transduced hMDSCs for regenerating functional bone in young and old mice. These findings correlated with lower phosphorylated p38MAPK expression and similar expression levels of cell survival genes and osteogenic-related genes in old hMDSCs relative to young hMDSCs. Old cells exhibited equivalent resistance to oxidative stress. However, both young and old donor cells regenerated less bone in old than young hosts. Impaired bone regeneration in older hosts was associated with high bone remodeling due to higher serum levels of RANKL and lower level of IGF-1. CONCLUSION: hMDSC-mediated bone regeneration was not impaired by donor age when hMDSCs were transduced with LBMP2/GFP, but the age of the host adversely affected hMDSC-mediated bone regeneration. Regardless of donor and host age, hMDSCs formed functional bone, suggesting a promising cell resource for bone regeneration.


Subject(s)
Aging , Bone Regeneration/physiology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/transplantation , Tissue Donors , Adult , Age Factors , Aged , Animals , Bone Morphogenetic Protein 2/genetics , Bone and Bones/injuries , Cell Differentiation/physiology , Disease Models, Animal , Female , Humans , Lentivirus , Male , Mice , Mice, Inbred ICR , Mice, SCID , Osteogenesis/physiology , Transduction, Genetic
9.
Stem Cells Transl Med ; 7(10): 705-708, 2018 10.
Article in English | MEDLINE | ID: mdl-30211981

ABSTRACT

We report the case of a patient who had undergone injections of myoblasts in an infarct area 16 years before being referred for heart transplantation. The pathological examination of the explanted heart found persisting myotubes embedded in fibrosis. This finding supports the ability of myoblasts to survive in harsh environments, which can make them appealing candidates for transplantation in diseases requiring supply of new myogenic cells. Stem Cells Translational Medicine 2018;7:705-708.


Subject(s)
Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , Adult , Fibrosis , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myosin Heavy Chains , Troponin T/metabolism , Ventricular Function, Left/physiology
11.
Cell Physiol Biochem ; 48(6): 2607-2655, 2018.
Article in English | MEDLINE | ID: mdl-30121644

ABSTRACT

A large number of clinical trials have shown stem cell therapy to be a promising therapeutic approach for the treatment of cardiovascular diseases. Since the first transplantation into human patients, several stem cell types have been applied in this field, including bone marrow derived stem cells, cardiac progenitors as well as embryonic stem cells and their derivatives. However, results obtained from clinical studies are inconsistent and stem cell-based improvement of heart performance and cardiac remodeling was found to be quite limited. In order to optimize stem cell efficiency, it is crucial to elucidate the underlying mechanisms mediating the beneficial effects of stem cell transplantation. Based on these mechanisms, researchers have developed different improvement strategies to boost the potency of stem cell repair and to generate the "next generation" of stem cell therapeutics. Moreover, since cardiovascular diseases are complex disorders including several disease patterns and pathologic mechanisms it may be difficult to provide a uniform therapeutic intervention for all subgroups of patients. Therefore, future strategies should aim at more personalized SC therapies in which individual disease parameters influence the selection of optimal cell type, dosage and delivery approach.


Subject(s)
Heart Diseases/therapy , Stem Cell Transplantation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Heart/physiology , Heart Diseases/pathology , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/transplantation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/transplantation , Regeneration , Ventricular Remodeling
12.
Tissue Cell ; 53: 93-103, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30060833

ABSTRACT

Mesenchymal stem cells (MSCs) are widely used in transplantation therapy due to their multilineage differentiation potential, abundance, and immuno-modulating ability. However, the risk of allograft rejection limits their application. Here, we proposed a novel method to facilitate MSC transplantation with enhanced applicability and efficacy. We cultured human adipose-derived MSCs in a 3D culture under in vitro expansion conditions and under conventional 2D adherent culture conditions. MSC spheroids promoted extracellular matrix molecules that stimulate MSC proliferation, and produced more angiogenic cytokines such as vascular endothelial growth factor, hepatocyte growth factor, and fibroblast growth factor than 2D-cultured MSCs. Further, MSC spheroids showed increased IDO expression, increased proportion of M2 macrophages, and decreased macrophage proliferation, compared to 2D-cultured MSCs. Next, we proposed the wrapping of autologous cell sheets from the recipient around in-vitro-grown MSC spheroids to prevent allogenic immune rejection during transplantation. Myoblasts from C57BL/6 mice were used to prepare a stem cell composite sheet containing human-derived MSC spheres. The transplantation of MSC spheroids increased the survival rate and decreased the inflammatory response of the immunocompetent C57BL/6 ischemic mice. Thus, combining 3D-cultured MSC spheroid technology with immune evasion stem cell composite sheet improved the outcome and strengthened the protection against allogenic immune rejection.


Subject(s)
Graft Rejection/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myoblasts, Skeletal/transplantation , Spheroids, Cellular/metabolism , Spheroids, Cellular/transplantation , Allografts , Animals , Autografts , Coculture Techniques , Humans , Mesenchymal Stem Cells/cytology , Mice , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Spheroids, Cellular/cytology
13.
Artif Cells Nanomed Biotechnol ; 46(sup3): S328-S335, 2018.
Article in English | MEDLINE | ID: mdl-30032651

ABSTRACT

A population of muscle-derived stem/progenitor cells (MDSPCs) contained in skeletal muscle is responsible for muscle regeneration. MDSPCs from mouse muscle have been shown to be capable of differentiating into pancreatic islet-like cells. However, the potency of MDSPCs to differentiate into functional islet-like cluster remains to be confirmed. The therapeutic potential of autologous MDSPCs transplantation on type 1 diabetes still remains unclear. Here, we investigated a four-stage method to induce the differentiation of MDSPCs into insulin-producing clusters in vitro, and tested the autologous transplantation to control type 1 diabetes in mice. MDSPCs isolated from the skeletal muscles of mice possessed the ability to form islet-like clusters through several stages of differentiation. The expressions of pancreatic progenitor-related genes, insulin, and islet-related genes were significantly upregulated in islet-like clusters determined by the quantitative reverse transcription polymerase chain reaction. The autologous islet-like clusters transplantation effectively improved hyperglycaemia and glucose intolerance and increased the survival rate in streptozotocin-induced diabetic mice without the use of immunosuppressants. Taken together, these results provide evidence that MDSPCs from murine muscle tissues are capable of differentiating into insulin-producing clusters, which possess insulin-producing ability in vitro and in vivo, and have the potential for autologous transplantation to control type 1 diabetes.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Myoblasts, Skeletal , Animals , Autografts , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred ICR , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Myoblasts, Skeletal/transplantation
15.
Interact Cardiovasc Thorac Surg ; 26(6): 993-1001, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29360997

ABSTRACT

OBJECTIVES: We previously reported that cell sheet transplantation combined with an omentopexy (OP) procedure is more effective for repairing heart damage when compared with cell sheet transplantation alone. However, a simultaneous (conventional) laparotomy as part of the OP may adversely affect the general condition of critically ill heart failure patients who would otherwise benefit from cell sheet transplantation, which is a paradox to be reconciled before this treatment can be applied in a clinical setting. We devised a novel endoscopic approach termed 'transphrenic peritoneoscopy' (TPP) for minimal access to abdominal organs from the thoracic cavity. Herein, we evaluated the feasibility and safety of TPP with an OP in a porcine myocardial infarction model. METHODS: Myocardial infarction was induced in 4 mini pigs by placing an ameroid constrictor around the left anterior descending artery. One month later, a left thoracotomy was performed in 2 randomly selected mini pigs, and a laparoscopic port was placed on the left diaphragm to gain access into the abdominal cavity. Using a low-pressure pneumoperitoneum, a flexible gastrointestinal endoscope was advanced, then the omentum was partially grasped with endoscopic forceps and brought back into the thoracic cavity via the diaphragm. Skeletal myoblast cell sheets were then implanted over the impaired myocardium, followed by placing the omentum over the sheets. RESULTS: TPP-assisted OP was accomplished in 2 post-myocardial infarction mini pigs with severe heart failure with an intra-abdominal pressure ≤8 mmHg within 30 min (22 and 27 min, respectively). Necropsy findings revealed a viable omentum flap and pedicle in both animals, with no evidence of procedure-related complications. Angiographic and histological analyses confirmed vessel communication between the omentum and the left ventricle. CONCLUSIONS: Our TPP approach was shown to be feasible and safe with a low-pressure pneumoperitoneum, while the omentum flap was durable. This successful combination of techniques may provide less-invasive endoscopic intervention and regenerative therapy.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Laparoscopy/methods , Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , Myocardium/pathology , Omentum/surgery , Animals , Coronary Angiography , Disease Models, Animal , Female , Heart Ventricles/pathology , Myocardial Infarction/diagnosis , Swine , Swine, Miniature
16.
Laryngoscope ; 128(3): 603-609, 2018 03.
Article in English | MEDLINE | ID: mdl-28842993

ABSTRACT

OBJECTIVE: There is an unmet need for tissue-engineered three-dimensional (3D) muscle constructs for laryngeal reconstruction. Functional engineered muscle could be used to repair postoncologic or traumatic defects or to medialize the vocal fold in cases of paresis/paralysis. Autologous, organized, engineered muscle that has adequate bulk integrates into host tissue and restores function currently does not exist. METHODS: Primary skeletal muscle progenitor cells (MPCs) were isolated from F344 rats. Three-dimensional muscle constructs were created by encapsulating MPCs via flow alignment in a customized collagen formulation and cultured under passive tension. Muscle-specific immunohistochemistry and confocal microscopy were used to evaluate muscle tissue differentiation. After 2 weeks of culture, muscle constructs were implanted into surgically created defects in the rat larynx. Postmortem function testing and histology was performed at 1 and 3 months. RESULTS: Immunohistochemistry with confocal microscopy demonstrated well-differentiated myotubes, which were well aligned and distributed throughout the engineered construct in vitro. There was evidence of restoration of normal laryngeal function at 1 month postoperative, as indicated by safe swallow (no aspiration events), weight gain, and excellent animal survival. Postmortem specimens demonstrated functional muscle contraction on ex vivo testing, and histology confirmed integration into host tissue. CONCLUSION: This is the first study to demonstrate that functional, 3D tissue-engineered skeletal muscle can be developed from primary MPCs and standardized oligomeric collagen. Collectively, these findings may have tremendous clinical implications for autologous laryngeal muscle repair and reconstruction. LEVEL OF EVIDENCE: NA. Laryngoscope, 128:603-609, 2018.


Subject(s)
Laryngoplasty/methods , Muscle, Skeletal/surgery , Myoblasts, Skeletal/transplantation , Tissue Engineering/methods , Animals , Collagen , Muscle Contraction , Muscle, Skeletal/cytology , Rats , Rats, Inbred F344
17.
Mol Ther ; 26(2): 618-633, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29221805

ABSTRACT

After intra-arterial delivery in the dystrophic dog, allogeneic muscle-derived stem cells, termed MuStem cells, contribute to long-term stabilization of the clinical status and preservation of the muscle regenerative process. However, it remains unknown whether the human counterpart could be identified, considering recent demonstrations of divergent features between species for several somatic stem cells. Here, we report that MuStem cells reside in human skeletal muscle and display a long-term ability to proliferate, allowing generation of a clinically relevant amount of cells. Cultured human MuStem (hMuStem) cells do not express hematopoietic, endothelial, or myo-endothelial cell markers and reproducibly correspond to a population of early myogenic-committed progenitors with a perivascular/mesenchymal phenotypic signature, revealing a blood vessel wall origin. Importantly, they exhibit both myogenesis in vitro and skeletal muscle regeneration after intramuscular delivery into immunodeficient host mice. Together, our findings provide new insights supporting the notion that hMuStem cells could represent an interesting therapeutic candidate for dystrophic patients.


Subject(s)
Muscle, Skeletal/physiology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/transplantation , Regeneration , Stem Cell Transplantation , Adult Stem Cells , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Mice , Muscle Development , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Regenerative Medicine
18.
Thorac Cardiovasc Surg ; 66(1): 42-52, 2018 01.
Article in English | MEDLINE | ID: mdl-29284167

ABSTRACT

Preclinical data suggested that somatic stem or progenitor cells derived induce and/or support endogenous repair mechanisms of the myocardium. Such cell populations were clearly shown to promote neovascularization in postischemic tissue, and some evidence also indicated transdifferentiation into cardiomyocytes. In the clinical setting, however, many attempts to regenerate damaged myocardium with a variety of autologous and allogeneic somatic progenitors have failed to generate the expected therapeutic efficacy. Currently, efforts are being made to select specific cellular subpopulations, modify somatic cells to augment their regenerative capacity, improve delivery methods, and develop markers selection of potentially responding patients. Cardiac surgical groups have pioneered and continue to advance the field of cellular therapies. While the initial excitement has subsided, the field has evolved into one of the pillars of surgical research and benefits from novel methods such as cellular reprogramming, genetic modification, and pluripotent stem cell technology. This review highlights developments and controversies in somatic cardiac cell therapy and provides a comprehensive overview of completed and ongoing clinical trials.


Subject(s)
Adult Stem Cells/transplantation , Bone Marrow Transplantation , Cardiovascular Diseases/surgery , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Myoblasts, Skeletal/transplantation , Myocardium/pathology , Regeneration , Regenerative Medicine/methods , Adult Stem Cells/metabolism , Animals , Bone Marrow Transplantation/adverse effects , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Myoblasts, Skeletal/metabolism , Myocardium/metabolism , Phenotype , Recovery of Function , Signal Transduction , Treatment Outcome
19.
Exp Toxicol Pathol ; 69(8): 598-604, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28583699

ABSTRACT

Although still a matter of controversial discussion, skeletal myoblasts are one of the options for stem cell transplantation improving cardiac function after myocardial infarction, exhibiting several advantages including the availability, the ability of self-renewal and differentiation, and the lack of ethical and immunological problems. The aim of this study was to investigate the impact of stem cell therapy with skeletal myoblasts on experimental venous bypass grafts in a mouse model of vein graft disease. Forty C57BL/6J mice underwent bypass grafting interposing a venous bypass graft of the donor mouse into the carotid artery of the recipient mouse. Twenty mice received periadventitially treatment with 1 million fluorescence labeled skeletal myoblasts suspended in culture medium (treatment group), the other twenty mice received only culture medium without myoblasts (control group). Two weeks after bypass surgery, the vein grafts of all 40 mice were harvested, stained and histologically investigated under light and immunofluorescence microscope. Against our expectations, skeletal myoblasts stayed in place and were still located in the adventitia after bypass grafting. Additionally, vein grafts of the myoblast group revealed a 2fold increased neoneointima formation, a decreased media thickness, a slightly increased neovascularization, a higher percentage of reendothelialization and also a slightly higher percentage of PDGFR ɑ, PDGFR ß, MMP-7 and MMP-9 positive cells, suggesting a paracrine mechanism responsible for accelerated neointima formation. In conclusion, the results of our study do not support the use of skeletal myoblast for the treatment of vein graft disease after coronary artery bypass surgery.


Subject(s)
Graft Occlusion, Vascular/prevention & control , Myoblasts, Skeletal/transplantation , Neointima/therapy , Stem Cell Transplantation , Animals , Carotid Artery, Common/surgery , Coronary Artery Bypass , Graft Occlusion, Vascular/pathology , Mice, Inbred C57BL , Treatment Outcome , Vena Cava, Inferior/transplantation
20.
Sci Rep ; 7(1): 3462, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615691

ABSTRACT

Satellite cells, localized within muscles in vivo, are Pax7+ muscle stem cells supporting skeletal muscle growth and regeneration. Unfortunately, their amplification in vitro, required for their therapeutic use, is associated with reduced regenerative potential. In the present study, we investigated if human myogenic reserve cells (MRC) obtained in vitro, represented a reliable cell source for muscle repair. For this purpose, primary human myoblasts were freshly isolated and expanded. After 2 days of differentiation, 62 ± 2.9% of the nuclei were localized in myotubes and 38 ± 2.9% in the mononucleated non-fusing MRC. Eighty percent of freshly isolated human MRC expressed a phenotype similar to human quiescent satellite cells (CD56+/Pax7+/MyoD-/Ki67- cells). Fourteen days and 21 days after cell transplantation in immunodeficient mice, live human cells were significantly more numerous and the percentage of Pax7+/human lamin A/C+ cells was 2 fold higher in muscles of animals injected with MRC compared to those injected with human myoblasts, despite that percentage of spectrin+ and lamin A/C+ human fibers in both groups MRC were similar. Taken together, these data provide evidence that MRC generated in vitro represent a promising source of cells for improving regeneration of injured skeletal muscles.


Subject(s)
Muscle Development , Regeneration , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism , Adult , Animals , Biomarkers , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Immunocompromised Host , Mice , Mice, Transgenic , Models, Animal , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation/methods , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...